The invention relates to two phase pharmaceutical compositions comprising as active ingredient a mao inhibitor and an uptake inhibitor together with usual pharmaceutical auxiliaries. The compositions can be used for the treatment of neurodegenerative diseases. As active ingredient optionally N-(1-phenyl-isopropyl)-N-methyl-propinylamine or N-(4-fluoro-phenyl)-isoprop-1-yl-N-methyl-propinylamine or their salts, optically active isomers or metabolites are used both as mao inhibitor and as uptake inhibitor.

Patent
   5589513
Priority
Dec 20 1991
Filed
Jun 17 1994
Issued
Dec 31 1996
Expiry
Dec 31 2013
Assg.orig
Entity
Large
31
4
EXPIRED
3. A transdermal pharmaceutical composition for treating a neurodegenerative disease which comprises:
(a) about 1.0 parts by weight of Carbowax 35 000;
(b) about 16.0 parts by weight of Carbowax 4000;
(c) about 53.0 parts by weight of Carbowax 400;
(d) about 2.0 parts by weight of 1,2-propylene-glycol;
(e) about 15.0 parts by weight of xanthan gum;
(f) about 7.0 parts by weight of deprenyl or p-fluorodeprenyl; and
(g) about 6.0 parts by weight of glycerine-polyethylene glycol-ricinoleate, said composition being in a liquid crystalline state containing translucent solid crystals of 8 to 10 microns.
1. A pharmaceutical composition for treating a neurodegenerative disease by selectively inhibiting mao-B activity without inhibiting mao-A activity and by inhibiting uptake of a biogenic amine to the brain, which consists essentially of:
(a) a fast-acting mao-B inhibitor in an amount effective to inhibit mao-B selected from the group consisting of deprenyl and p-fluoro-deprenyl or a pharmaceutically acceptable acid addition salt thereof; and
(b) a slow-acting inhibitor of uptake of a biogenic amine in an amount effective to inhibit uptake of a biogenic amine, said slow-acting inhibitor of uptake of a biogenic amine selected from the group consisting of deprenyl and p-fluoro-deprenyl or a pharmaceutically acceptable acid addition salt thereof.
2. The pharmaceutical composition defined in claim 1 wherein the fast-acting mao-B inhibitor contains deprenyl or a pharmaceutically acceptable acid addition salt thereof and the slow-acting inhibitor of a biogenic amine contains deprenyl or a pharmaceutically acceptable acid addition salt thereof.

This is a 371 of PCT/Hu92/08060, filed 12/18/92.

The invention relates to a pharmaceutical composition, suitable for the treatment of neurodegenerative diseases, containing active ingredients, resulting in adequate medicinal concentrations in time and level, both in the blood and the brain, and to a process for the preparation thereof.

It is known, that monoamine oxidase (MAO) is one of the main metabolizing enzyme [Blaschko H. Pharmacol. Rev. 4, 415, (1951)] of biogenic amines occurring in the human nerve cells. Due to its activity the biogenic amines, playing an essential role in neurotransmission, are decomposed into ineffective metabolites. It was recognized that in certain diseases the level of the brain biogenic amines was decreased.

Agents, which inhibit the metabolizing enzyme (enzymes) can restore the normal level of these amines. This is the reason why MAO inhibitors were introduced in the human therapy. There are observations that MAO-inhibitions may lead to a serious side effect which is connected to tyramine (structurally a biogenic amine) potentiation ("cheese reaction") which is derived from foods and may induce blood pressure increase, and can be lethal. [Piackar and co-workers, Psychopharmacology 73, 3087, (1981)].

MAO exists in two forms, termed MAO-A and MAO-B. Inhibiting the B form selectively, the A form is able to decompose tyramine, which is a mixed type of substrate and the dangerous side effect can be avoided. This selective MAO inhibition can be accomplished by administration of (-)-deprenyl, [(-)N-(-1-phenyl-isopropyl)-N-methyl-propinylamine-hydrochloride)] which selectively and irreversibly inhibits the MAO-B enzyme [Elsworth et al., Psychopharmacology 57, 33, (1978)]. Because of the irreversible inhibition, the recovery of the enzyme activity can only be due to new enzyme resynthesis.

The development of the irreversible inhibition of the enzyme involves two steps. The first one is reversible and only the formation of the second enzyme-inhibitor complex becomes irreversible. The half life of enzyme regeneration is 7-8 days. [Oreland et al., J. Neural. Transm. Suppl. 32, 55-59 (1990)].

The substrate specificities of the enzymes and the selectivity of the mostly known inhibitors are reviewed by Dostert and his co-workers. [Medicinal Research Reviews, Vol 9, No. 1. 45-89, (1989)].

Balard [Science 219, 979-980, (1980)] and Burns (Proc. Natl. Acad. Sci 80, 4546-4550 (1983)] described that MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) due to its neurotoxic activity evokes parkinsonian syndrome in man and similar symptoms can be observed in animal experiments. The MPTP causes selective damage of the dopaminergic neurons to the corpus striatum. The histological alterations are similar to those, observed in postmortem parkinsonian brains. It is known, that this effect of MPTP can be prevented with MAO inhibitors, especially with deprenyl. The preventing role of (-)-deprenyl is due to the inhibition of the conversion of MPTP to MPP+, [Nature, 311, 467, (1984)]. The MPTP induced neuronal damage can be retarded also with dopamine uptake inhibitors [Proc. Natl. Acad. Sci. USA, 82, 2175, 1985] like mazindol, by inhibiting the active uptake of MPP+ (methyl-phenyl-pyridinium ion) into the dopaminergic neurons.

During the period of MAO activity hydrogen peroxide and oxygen free radicals are formed, which can lead to oxydative damage of the neurons. Ammonia and some heterocyclic isoquinolines can also be formed by the MAO which can be considered neurotoxic. [Maret et al., Drug metabolism. Reviews, 22, 291-332, (1990); P. Riederer et al., Acta Neurol. Scand. 126, 41 (1989); Benedetti and Dostert, Biochem. Pharm. Vol 38, 555, (1989)].

It is known that DSP-4 [N-(2-chloroethyl)-N-ethyl-2-bromo-benzylamine], a neurotoxic agent, induces noradrenaline (NA) depletion selectively from the central and peripheral noradrenergic neurons [Grzanna et al., J. Histochem. Cytochem., 1435-1442, (1989)].

It is further known that--reuptake inhibitors such as desipramine [10,11-Dihydro-N-methyl-5-H-dibenz(6,7)azepine-5-propanamine) inhibit the NA-depleting effect of DSP-4 [Johnsson et al., Neuroscience, 7, 2895, (1982); Ross Br. J. Pharmacol., 58, 521, (1976)]--MDL 72974A [(E)-4-fluoro-beta-fluoro-ethylene benzene butamine] a highly selective MAO-B inhibitor lacks catecholaminergic reuptake blocking properties and fails to prevent DSP-4 induced toxicity [Finnegen et al. Eru. J. of Pharmacol., 184, 119-126 (1990)].

It became clear, that (-)-deprenyl cannot be considered only as a simple selective, irreversible inhibitor of MAO-B. It was stated, that it inhibits the uptake of dopamine, noradrenaline and tyramine in the nerve terminals and into the peripheral ganglions, but only in extremely high doses [Knoll, Advances in Biochem. Psychopharmacology Vol, 5, 393,(1972)]. It must be kept in mind that, in addition to the MAO inhibitory effect deprenyl possesses an uptake inhibitory activity.

The object of the invention is to prepare a pharmaceutical composition with optimal properties for the treatment of neurodegenerative diseases.

In the course of our experiments we learned that

1.) Longlasting MAO inhibition can be achieved only when the concentration of the inhibitor in the brain and blood reaches a sufficiently high (15-40 pmol/mg tissue) concentration. When the concentration of the inhibitor is too high (30 mg/die) the concentration of the metabolites will also be high enough to cause an undesired psychostimulant effect and/or the selectivity of the inhibitor will also be lost (MAO-A is also inhibited).

2.) Deprenyl and p-fluoro-deprenyl [N-(4-fluoro-phenyl)-isoprop-1-yl]-N-methyl-propinyl-amine exert their activity both as the parent (unchanged) compound and as the metabolites.

The results are illustrated in FIGS. 1 and 2.

After oral administration of alternatively and positionally labelled deprenyl and p-fluoro-deprenyl (ring-3 H and propargyl-14 C (1.5, 10 mg/kg, respectively) the distribution of the compounds were tested in 15 brain regions (the paired brain parts were separtely studied all together in 25 brain areas and in the plasma during 96 h, as a function of time in rats. It was stated, that the unchanged compound was quickly absorbed (15 minutes), and penetrated into the central nervous system. The unchanged deprenyl molecule has a short residence time in the brain, but the metabolites can be found for a longer period, in the tissues.

The simultaneous presence and the equal quantities of the two labels indicate the unchanged molecule (the data relate to the molar concentrations calculated on the basis of the two labels). Because of the fast change observed in the injected 3 H/14 C ratio in the tissues, our experiments indicate a significant metabolite formation (amphetamine, methylamphetamine, p-fluoro-methylamphetamine and p-fluoro-amphetamine) and their presence in the brain.

3.) The potential metabolites of deprenyl and p-fluoro-deprenyl, (methylamphetamine, amphetamine and p-fluoro-methylamphetamine, p-fluoro-amphetamine, respectively) have a significant uptake inhibitory potency; they can prevent in vivo the MPTP neurotoxicity without the development of a high degree of MAO inhibition. The results are shown in tables 1, 2 and 3. The tests were carried out according to the method of Heikilla [Nature, 311, 467-469 (1984)].

4.) The potential metabolites of deprenyl and p-fluoro-deprenyl (methylamphetamine, amphetamine and p-fluoro-methylamphetamine, p-fluoro-amphetamine, respectively) are able to prevent in vivo in a dose of 1-5 mg/kg (i.p.) the DSP-4 induced neurotoxicity without the development of MAO inhibition.

The results are shown in Table 8.

The tests were carried out according to the method of Finnegan [Finnegan et al., Eur. J. Pharmacol., 184, 119-126, (1990)].

Using the metabolites in high concentration in vivo (10 mg/kg; i.p.) they potentiated the toxic effect of the DSP-4 which was indicated by the death of the animals.

5.) To prevent the neurotoxicity either a properly high concentration of the Unchanged compound for a shorter period (a concentration necessary to complete irreversible MAO-B inhibition) or because of the reversibility of the uptake inhibition, the longlasting presence of the metabolites are needed.

It was found that maximal effect can be reached when both conditions are fulfilled.

FIG. 1 is a bar graph showing the level of deprenyl labelled with 3 H on the phenyl moiety and 14 C on the propargyl moiety in 25 different areas of a rat brain 45 minutes after oral administration of the labelled deprenyl (1.5 mg/kg) to the rat based on measurement of the 3 H and 14 C.

FIG. 2 is a bar graph showing the level of p-fluoro-deprenyl (CH-175) labelled with 3 H on the phenyl moiety and 14 C on the propargyl moiety in 25 different areas of a rat brain 45 minutes after oral administration of the labelled p-fluoro-deprenyl (10 mg/kg) to the rat based on measurement of the 3 H and 14 C.

FIG. 3 is a series of 5 graphs plotting the plasma level of amphetamine in pigs, expressed in ng/ml along the Y-axis, against time expressed in hours along the X-=axis. The amphetamine in the plasma in each case is a metabolite resulting from administration to the pig of Selegiline, which is (-)-deprenyl, either orally, intravenously, or orally in a two phase composition containing the drug in both fast release and slow release form in various proportions.

FIG. 4 is a series of 2 graphs plotting along the Y-axis the level of noradrenaline (NA) in the hippocampus of rats against time, expressed in hours along the X-axis. The rats in each case were orally administered 3 mg/kg of (-)-deprenyl. In one case the entire 3 mg/kg were administered as a "fast" composition, in the other case the 3 mg/kg were administered as 1 mg/kg "fast" and 2 mg/kg "slow". Following oral administration of (-)-deprenyl, by 1,2,4 or 8 hours, DSP-4 was administered to the rats, intraperitoneally, and the inhibition of DSP-4 neurotoxicity was comparatively determined in each case.

FIG. 5 is a series of two graphs plotting survival rate of rats as a percentage along the Y-axis, against time, expressed in hours along the X-axis. In each case the total amount of (-)-deprenyl orally administered is 4 mg/kg as either all "fast" composition or as 2 mg/kg "fast" and 2 mg/kg "slow". Once again DFP-4 was intraperitoneally administered to the rats after the (-)-deprenyl was administered.

The invention relates to two phase pharmaceutical compositions comprising as active ingredient a compound with MAO inhibiting effect and a compound with uptake inhibiting effect together with usual auxiliary materials.

The compositions according to the invention contain as active ingredient 5-95 mass % of a reversible or irreversible MAO inhibitor, 5-95 mass % of an uptake inhibitor advantageously of sustained activity in a ratio of 1-19: :19-1, advantageously in a ratio of 1:1, 1:2 or 1.3.

The composition is administered depending on the state of the patient, on the seriousness of the clinical picture and on the individual sensitivity of the patient in a dose of 5-20, advantageously 10 mg/day.

If the uptake inhibitor is not of a sustained activity, the inhibitor is advantageously used in a retard form.

As MAO inhibitor advantageously deprenyl, p-fluoro-deprenyl, their salts, or optically active isomers respectively can be applied.

As uptake inhibitors advantageously deprenyl, p-fluoro-deprenyl or a compound of the formula I ##STR1## can be used, wherein R1 is straight or branched C1-8 alkyl, C7-10 phenylalkyl, phenyl, C3-8 cycloalkyl, R2 is a straight or branched C1-8 alkyl, a C1-8 alkyl substituted by a halogen atom, by hydroxy-C1-4 alkoxy or by one or two phenyls, phenyl or C3-8 cycloalkyl with the proviso, that R1 and R2 contain together at least 3 carbon atoms--or an acid addition salt, or metabolite thereof.

As a compound of the formula I advantageously N-propyl-1-phenyl-2-pentylamine or its acid addition salt, N-propyl-1-phenyl-2-butylamine or its acid addition salt or N-propyl-1-phenyl-2-hexylamine or its acid addition salt can be used.

The two phase compositions according to the invention can be prepared by methods known per se in the form of pellets, tablets, coated tablets, transdermal compositions, dragee, suspension containing microcapsules, capsules, coated capsules, per os suspensions, suspension injections using known auxiliaries.

As auxuliaries advantageously the following materials can be used:

a.) As filling materials: saccharose, lactose, mannitol, starch, starch-talc-saccharose, calcium phosphate, hydroalcoholic solution of polyvidone (polyvinylpyrrolidone), etc. can be used.

b.) auxiliaries:

lypophilic base (stearic acid, stearic acid palmitate, glyceryl-ditripalmite-stearate, etc.);

Other auxiliaries known in the pharmaceutical technology: Eudragit derivatives, etc.

Cellulose-derivatives: HPMC, CMC, EC and their salts.

c.) Granulating liquid: water, ethanol, ethanol-water, isopropanol, isopropanol-water.

d.) Binders: PVP/VA, Eudragit derivatives, cellulose derivatives, and salts thereof.

One can proceed in a way too, that the pharmaceutical composition is prepared in situ, that is the patient is treated simultaneously with a suitable dose of MAO inhibitor needed to reach continuously a 98% MAO inhibition and a suitable dose of uptake inhibitor needed to reach a continuous uptake inhibition.

The preparation of deprenyl, p-fluoro-deprenyl, and compounds of the formula I is described in the U.S. Pat. No. 4,564,706; European patent specification No. 186,680 and Portuguese patent specification No. 85.799.

PAC 1.) A two phase tablet is prepared by a method known per se:

Composition

Internal phase: prepared by double granulation

______________________________________
a.) Deprenyl: 5 mg
Methocel k 4M premium:
50 mg
Lactose: 20 mg
______________________________________

Quantum satis of isopropanol for the preparation of granules.

______________________________________
b.) Deprenyl: 10 mg
Amylum maydis:
40 mg
Avicel PH-101:
20 mg
PVP K-25: 10 mg
______________________________________

Quantum satis of isopropanol for the preparation of granules (also distilled water can be used).

______________________________________
External phase:
______________________________________
Mg stearin: 8 mg
Talcum: 15 mg
Aerosil-200: 2 mg.
______________________________________
______________________________________
Component
______________________________________
Deprenyl 15 mg
Stearic acid 30 mg
Eudragit RSPM 45 mg
Collidon VA 64 47 mg
Carbopol 940 35 mg
Sterotex 25 mg
Magnesium stearate
3 mg
220 mg
______________________________________
(Diameter of tablets 8 mm)

1. Preparation of granules by dry granulating method well-known per se.

The sequence of powder-mixing: Collidon VA 64, Carbopol 940, Stearic acid, sterotex (atomized) (hydrogenated cottonseed oil), Eudragit RSPM (acrylic-methacrylic acid ester copolymer), Deprenyl.

2. Tabletting of powder mixture by eccentric tabletting machine at low r.p.m.

Results of dissolution test. Three parallel measurements were made. One cell contained 15 tablets.

a.) 316.5 cg/15 pieces

b.) 316.0 cg/15 pieces

c.) 319.0 cg/15 pieces

The medium used for dissolution was artificial gastric fluid without pepsin. (Ph. Hg. VII.)

Dilution is zero

d=1 cm

λ=256 mm

______________________________________
Results
Time (minute)
Dissolved active substance (%)
______________________________________
10 4.5
15 9.0
30 21.0
60 31.2
90 47.7
120 39.3
150 41.1
180 48.6
240 51.9
300 63.3
360 69.0
420 (7 hours)
75.6
______________________________________

3.) preparation of two phase pellets--"fast" and "slow" pellets with the following compositions are prepared.

______________________________________
* FAST PELLETS
Neutral pellets (sugar spheres)
61.1 mg
Deprenyl 16.3 mg
Lactose 16.6 mg
Polyvidone (K30) 6.3 mg
* SLOW PELLETS
Neutral pellets (sugar spheres)
44.6 mg
Deprenyl 11.9 mg
Lactose 11.9 mg
Polyvidone (K30) 4.6 mg
Ethylcellulose about 8.4 mg
Talc about 16.9 mg
Castor oil about 1.7 mg
manufacturing process
______________________________________

Saccharose crystals are coated with a mixture of starch-saccharose and a hydroalcoholic solution of polyvidone and saccharose (sugar sphere).

A mixture of Deprenyl hydrochloride and lactose is attached on the microgranules using an alcoholic solution of polyvidone.

In the case of the "slow" pellet on the microgranules obtained an alcoholic varnish of ethylcellulose (plastified with Castor oil) and talc is applied.

To obtain the required "fast and slow" pellet the desired amounts of the "fast" and the "slow" pellets are mixed in a capsule by a method known per se.

______________________________________
Components:
______________________________________
Carbowax 35000 1.0
Carbowax 4000 16.0
Carbowax 400 53.0
1,2-propylene-glycol
2.0
Xanthan gum 15.0
Deprenyl 7.0
Cremophor EL 6.0
100.0
______________________________________

Carbowax components were fused and the mixture was poured into mixing vessel of Erweka type at 50°C and was mixed on stage 6. Deprenyl was dissolved and suspended in a mixture of propylene glycol and Cremophor EL at 50°C and the mixture in portions (50 g each) was added into the mixing vessel. After each portion the mixture was mixed 1 minute. Finally at increased speed (stage 9) Xanthan gum was added in portions (5 g each). After each portion the mixture was mixed 1 minute. Thereafter the speed of mixing was decreased to stage 3.5 and mixing was continued until cooling (appr. 1.5 hours). Content of active substance is 6.84% and stability is appropriate.

Liquid crystalline state: 100%, with translucent solid crystals of 8-10 microns. (Cremophor EL: glycerin-poly-ethylenglycol-ricinoleate. Xanthan gum: polysaccharide).

Planning the experimental conditions the following data have to be considered.

Direct in vivo measurement of the uptake inhibitory potency of the metabolites was not possible, because of their reversible nature.

The MAO-B inhibitor potency--using the platelet MAO-B activity as an indicator--peripherally is measurable in pigs (the platelets originated from other species do not contain good measurable MAO-B enzyme activity).

Measuring the time dependency of DSP-4 neurotoxicity in rats (using the noradrenaline content of hippocampus as an indicator) after pretreatment with different dosage forms of deprenyl the correlation between the lack of neurotoxicity of DSP-4 and the adequate but not extremely high blood level of the metabolites of deprenyl, can be demonstrated.

In a series of experiments the permanent level of deprenyl metabolites in the blood of the domestic pigs was measured with analytical techniques, and the platelet, brain and liver MAO activity was also determined.

The experiments were carried out on female (bit white) domestic pigs, weighing 20-25 kg. The pigs were caged separately during the experiments, and the same food was supplied, which was used formerly.

Animals were treated orally with 5; 7.5; 10; 15 mg of (-)-deprenyl in a pellet and with 10 mg iv. Blood samples (5 ml) were taken at 0; 0.08; 0.25; 0.5; 0.75; 1; 1.5; 2; 3; 4; 6; 12; 24 and 48 h in centrifuge tubes containing 500 IU heparin for analytical measurement. The samples were centrifuged at 1500 r.p.m. for 10 min. to separate plasma. The metabolites were determined by GC method, using Hp-5890 gas chromatograph, eluation time for methylamphetamine: 21.4 min.

The results are shown in FIG. 3.

The inhibition of DSP-4 neurotoxicity was measured in rats. For treating rats the schedule of Finnegan was followed [Finnegan et al., Eur. J. Pharmacol., 184, 119-126, (1990)].

Wistar male rats, weighing 170-200 g were used. Animals were housed six to a cage under constant temperature (22°C), in a room illuminated 12 h per day. Food and water were freely available. Rats were pretreated orally with 3 mg/kg of the combination of example 3 (1 mg/kg "fast" (F)+2 mg/kg "slow" (S)), 1, 2, 4 and 8 hours before DSP-4 treatment (50 mg/kg; intraperitoneally).

Before the (-)-deprenyl pretreatment, the animals were starved for 24 h. Each hippocampus of the rats was by HPLC technique.

Results are shown in FIG. 4.

As it can be seen from FIG. 4, 3 mg/kg deprenyl p.o. can prevent the neurotoxicity of 50 mg/kg DSP-4 ip. in rats. In the case of the control ("fast") formulation this prevention is in the first period high (86.1%) but it decreases exponentially and after 8 hours reaches the level of DSP-4 treated control (19,6%). The claimed formulation produces 58.8% of inhibition, the inhibition is practically identical (54.5%) with that of the control formulation (51,5%) after two hours, but is significantly higher (43.31%) than that of those (16%) and does practically not change over eight hours. In the case of the treatment with 4 mg/kg deprenyl we found that the control ("fast") formulation was facilitated the neurotoxicity of DSP-4 time dependently in contrast to the claimed ("fast and slow") formulation. (See FIG. 5) This means that the dose of the "fast" component can not be increased unlimitedly without appearance of unwanted side effects.

Oppositely, applying the claimed compositions with increasing amounts of the "slow" part the inhibition of MAO-A will increase which means that the selectivity of deprenyl will be lost.

The results are given in Table 4.

TABLE 4
______________________________________
Percentage inhibition of MAO by deprenyl preparations
Brain
Preparation MAO-B MAO-A
______________________________________
1 mg/kg (tablets)
88.02 ± 1.49
2.55 ± 0.56
0.5 ± 0.25 mg/kg
85.84 ± 4.16
14.83 ± 3.37
("fast" and "slow")
0.5 ± 1.0 mg/kg
90.50 ± 3.10
27.28 ± 3.93
("fast" and "slow")
______________________________________

In the case of transdermal preparations the control group was treated orally with 10 mg of (-)-deprenyl in a gelatinous capsule.

Blood samples were taken for the determination of MAO-B activity at: 0, 3, 6, 24, 48, 72 and 96 h. At 96 h after blood sampling the pigs were killed and the MAO-B and MAO-A activity were determined in their dissected brain.

The second group was treated as control with the UG-111 transdermal preparation containing 10 mg (-)-deprenyl. The times of blood sampling were at: 0, 3, 6, 24 and 48 h. The transdermal preparations were removed at 24 h. For the determinations of the residual (-)-deprenyl content of the preparations the patch and its nylon cover were used. The skin was washed with ethanolic cotton-wool, which was also used for HPLC determination. The pigs were killed at 48 h and MAO-A and MAO-B activity of the brain was determined.

The third group of the pigs was treated as control with UG-167 containing 20 mg of (-)-deprenyl. Blood samples were taken at: 0, 3, 6, 24, 48 and 72 h. The patches were removed at 48 h and the whole procedure described at group 2 was accomplished.

The fourth group of pigs was treated with UG-191 containing 30 mg of (-)-deprenyl. Blood samples were taken at: 0, 3, 6, 24, 48, 72 and 96 h. The patches were removed at 48 h and the whole procedure described at group 2 was accomplished.

The blood was taken from the v. cava cranialis with a 20 ml plastic syringe containing 1.5 ml of 7.6% Na-citrate solution. The volume of the blood taken was 18.5 ml at every sampling.

MAO activity was measured radiometrically according to the methods of Wurtman and Axelrod [Biochem. Pharmacol. 12. 1414-19; (1963)] with a slight modification [K. Magyar in: Monoamine Oxidases and their Selective Inhibition, Ed.: K. Magyar, Pergamon Press, Akademiai Kiado, Budapest, 11-21; (1980)].

The method described by Willberg and Oreland was followed for platelet preparation. (Med. Biol., 54: 137-44; 1976).

TABLE 5
______________________________________
Absorption of (-)-deprenyl form transdermal preparations
as a function of time in pigs. (The residual (-)-deprenyl
content of the patches was determined with HPLC
technique).
Transdermal Duration of Remaining Deprenyl
preparation experiment (h)
% + S.D.
______________________________________
UG-111 24 14.2 ± 5.5
(control
10 mg
UG-167 24 36.5 ± 9.3
(control)
20 mg 6.1 ± 5.1
UG-191 24 58.5 ± 14.1
20 mg 48 6.1 ± 5.1
72 8.4 ± 2.4
______________________________________

The results of the inhibition of MAO-B activity of the platelet after p. os and transdermal application are shown in Table 6.

TABLE 6
__________________________________________________________________________
Effect of (-)-Deprenyl on the inhibition of MAO-B activity
of the platelets (%) as compared to the control. Measure-
ments vere made with 14 C-PEA substrate. ±B.D. (n = 3)
Mode of
time
application
3 6 24 48 72 96
__________________________________________________________________________
97.77 86.04 100.00 82.67 72.32 0.0
92.52 96.51 100.00 70.48 65.63 0.0
per os 0.0 95.63 100.00 69.93 50.17 0.0
(10 mg)
95.15 92.73 ± 3.35
100 ± 0.0
74.36 ± 4.16
52.71 ± 6.56
--
UG-111 0.0 36.27 52.68 60.70 -- --
(control)
trans- 54.80 86.47 86.02 92.03 -- --
dermal 95.66 95.47 93.76 98.63 -- --
(10 mg, 24 h)
75.23 72.74 ± 18.42
77.49 ± 12.6
83.79 ± 11.7
-- --
UG-167 23.29 55.77 90.94 88.56 100.00 --
(control)
trans- 72.11 95.90 98.54 91.44 89.34 --
dermal 65.81 65.05 0.0 90.57 75.02 --
(20 mg, 48 h)
53.74 ± 15.33
72.24 ± 12.13
94.74 90.19 ± 0.85
88.12 ± 7.24
--
UG-191 85.76 90.55 98.18 94.02 95.08 96.52
transderm.
79.83 93.32 95.73 94.66 95.87 96.16
(30 mg, 72 h)
0.0 0.0 67.75 95.51 94.22 92.64
82.80 91.94 87.22 ± 9.76
94.73 ± 0.43
95.06 95.17
__________________________________________________________________________

The results of the determination of MAO activity in the brain are shown in Table 7.

TABLE 7
__________________________________________________________________________
Effect of (-)-Deprenyl on the inhibition of MAO activity
(%) an compared to the control. Measurements were made in
domestic pig brain nucleous free homogenates and in liver
with 14 C-PEA; 14 C-S-HT substrate. ±B.S. (n = 3)
brain liver
Mode of application
14 C-PEA
14 C-5-HT
14 C-PEA
14 C-5-HT
__________________________________________________________________________
p. os (96 h)
73.22 ± 8.13
20.14 ± 6.0
41.04 ± 7.62
10.45 ± 6.4
UG-111
(48 h)
transdermal
(24 h)
56.31 ± 10.03
16.39 ± 8.77
28.80 ± 5.15
13.7
0
0
UG-167
(72 h)
86.76 ± 6.67
18.50 ± 3.81
30.30 ± 10.23
19.4
transdermal
(48 h)
86.76 ± 6.67
18.50 ± 3.81
30.30 ± 10.23
19.4
0
0
UG-191
(96 h)
transdermal
(72 h)
88.82 ± 1.21
15.52 ± 3.35
16.92 ± 10.59
0
__________________________________________________________________________

The composition, particle size, the precentage of liquid crystalline state of the different control preparations was as follows:

______________________________________
UG-111
______________________________________
PEG 4000 16.0 g
PEG 400 60.0 g
Propyleneglycol 8.0 g
Cremophor EL 2.0 g
Deprenyl HCL 5.0 g
PEG 400 ad 100.0 g
______________________________________

Average particle size: 72.7 microns; liquid crystalline state: 20%.

______________________________________
UG-167
______________________________________
PEG 4000 19.0 g
PEG 400 55.0 g
Propyleneglycol 8.0 g
Xanthan gum 10.0 g
Deprenyl HCl 5.0 g
PEG 400 ad 100.0 g
______________________________________

Average particle size: 91-109 microns; liquid crystalline state: 70-80%.

TABLE 1
______________________________________
IC50 values of the Deprenyl and PFD metabolites on the
MPP+ uptake to the striatatal synaptosones.
IC50 IC50
Compound M Compound M
______________________________________
(-)-deprenyl
-- (-)-p-F-deprenyl --
(-)-desmethyl-
-- (-)-p-F-desmethyl-
--
deprenyl deprenyl
(+)-desmethyl-
9.1 ×
deprenyl 10-6
(-)-metamphet-
4.9 ×
(-)-p-F-metamphetamine
1.3 × 10-6
amine 10-7
(+)-metamphet-
9.1 ×
(+)-p-F-metamphetamine
1.3 × 10-6
amine 10-8
(-)-p-F-amphetamine
7.2 × 10-7
(+)-p-F-amphetamine
1.0 × 10-7
Homifensin
3.1 ×
10-7
Mazindol 1.7 ×
10-7
______________________________________
TABLE 2
__________________________________________________________________________
In vitro uptake inhibition on rat brain
synaptosomas
NA DA 5-HT
COMPOUND HYPOTHALAMUS
STRIATUM
HIPPOCAMPUS
__________________________________________________________________________
(-)-DEPR. 5.1 × 10-5
1.0 × 10-4
5.0 × 10-3
(+)-DEPR. 1.7 × 10-5
2.4 × 10-5
3.6 × 10-2
(-)-p-FLUORO-D
1.3 × 10-5
2.9 × 10-5
1.4 × 10-3
(+)-p-FLUORO-D
6.1 × 10-6
1.6 × 10-5
6.0 × 10-4
(-)-METHYLAMPH.
3.5 × 10-6
4.2 × 10-5
--
(+)-METHYLAMPH.
3.5 × 10-7
6.0 × 10-7
1.9 × 10-2
(-)-p-FLUORO-MA
7.5 × 10-6
3.0 × 10-4
--
(+)-p-FLUORO-MA
7.7 × 10-6
2.3 × 10-5
1.7 × 10-3
__________________________________________________________________________
TABLE 3
__________________________________________________________________________
The effect of deprenyl posttreatment on MPTP neurotoxicty
1. time beetwen
2.
treatment
treatments 30
treatment
Dopamine
% DO
% MVA %
__________________________________________________________________________
physiological salt
phys.s.
15.1 ± 0.4
100
0.9 ± 0.02
100
1.4 ± 0.06
100
MPTP " 8.0 ± 0.5a
53 0.7 ± 0.04a
77
1.3 ± 0.07a
79
MPTP Deprenyl
15.3 ± 0.4b
101
0.9 ± 0.04b
100
1.3 ± 0.05c
93
MPTP Pargyline
7.1 ± 0.6NS
47 0.5 ± 0.04NS
55
0.9 ± 0.09NS
64
Pargyline MPTP 13.8 ± 0.5b
91 1.0 ± 0.06b
111
1.4 ± 0.06b
100
MPTP Mazindol
14.0 ± 0.8b
98 1.1 ± 0.08b
122
1.7 ± 0.10b
121
__________________________________________________________________________
a significantly lower than the control (p 0.01)
b significantly higher than group treated with MPTP (p. 0.01)
c significantly higher than group treated with MPTP (0.05 p 0.01)
NS no significant
DOPAC and HVA: metabolites of dopamine
TABLE 8
______________________________________
Interaction of (-)-deprenyl and (-)-p-fluoro-deprenyl me-
tabolites with DSP-4 neurotoxicity in rats
Na content of
Pretreatment
Treatment hippocampus
mg/kg; i.p. DSP-4 in % ± S.D.
Survival %
______________________________________
(-)-MA 1 50 61 ± 3.5
100
(-)-MA 5 30 116 ± 2.1
63
(-)-MA 10 50 nd* 0
(-)-p-F-MA
10 50 nd* 0
______________________________________
*nd: not determined. Because of the toxic interaction animals died within
the first day.
MA = methylamphetamine
pF-MA = pfluoro-methylamphetamine.

Marmarosi, Katalin, Szatmari, Istvan, Hermecz, Istvan, Kormoczy, Peter, Sziraki, Istvan, Torok, Zoltan, Gaal, Jozsef, Lengyel, Jozsef, Magyar, Kalman, Szabo, Anna Z.

Patent Priority Assignee Title
10226473, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
10561615, Dec 10 2010 Lipocine Inc. Testosterone undecanoate compositions
10653706, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
10716794, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
10799513, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
10881671, Nov 30 2010 Lipocine Inc High-strength testosterone undecanoate compositions
10973833, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
11052096, Jan 08 2009 Lipocine Inc. Steroidal compositions
11298365, Aug 28 2014 Lipocine Inc Bioavailable solid state (17-β)-hydroxy-4-androsten-3-one esters
11304960, Jan 08 2009 LIPOCINE, INC Steroidal compositions
11311555, Nov 30 2010 Lipocine Inc High-strength testosterone undecanoate compositions
11364249, Nov 30 2010 Lipocine Inc High-strength testosterone undecanoate compositions
11364250, Nov 30 2010 Lipocine Inc High-strength testosterone undecanoate compositions
11433083, Nov 30 2010 Lipocine Inc High-strength testosterone undecanoate compositions
11559530, Nov 28 2016 Lipocine Inc Oral testosterone undecanoate therapy
11707467, Aug 28 2014 Lipocine Inc (17-ß)-3-oxoandrost-4-en-17YL tridecanoate compositions and methods of their preparation and use
11872235, Aug 28 2014 LIPOCINE, INC Bioavailable solid state (17-β)-Hydroxy-4-Androsten-3-one esters
5847216, Sep 13 1994 KNOLL Aktiengesellschaft Preparation of selegiline
6391914, Oct 29 1998 FUJIMOTO CO , LTD Optically active aminopentane derivatives
7374779, Feb 26 1999 LIPOCINE, INC Pharmaceutical formulations and systems for improved absorption and multistage release of active agents
8778922, Jan 08 2009 Lipocine Inc. Steroidal compositions
8865695, Jan 08 2009 Lipocine Inc Steroidal compositions
9034858, Nov 30 2010 Lipocine Inc.; Lipocine Inc High-strength testosterone undecanoate compositions
9205057, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
9358241, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
9480690, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
9498485, Aug 28 2014 Lipocine Inc Bioavailable solid state (17-β)-hydroxy-4-androsten-3-one esters
9757389, Aug 28 2014 Lipocine Inc Bioavailable solid state (17-β)-hydroxy-4-androsten-3-one esters
9757390, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
9943527, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
9949985, Nov 30 2010 Lipocine Inc. High-strength testosterone undecanoate compositions
Patent Priority Assignee Title
5326770, Jul 17 1992 Bristol-Myers Squibb Pharma Company Monoamine oxidase-B (MAO-B) inhibitory 5-substituted 2,4-thiazolidinediones useful in treating memory disorders of mammals
5380761, Apr 15 1991 Chinoin Gyogyszer- es Vegyeszeti Termekek Gyara Rt. Transdermal compositions
5444095, Apr 04 1991 GOVERNING COUNCIL OF THE UNIVERSITY OF TORONTO, THE Use of deprenyl to rescue damaged nerve cells
5508311, Mar 01 1991 University of Saskatchewan Aliphatic propargylamines as selective MAO-B inhibitors and as neuroprotective agents
///////////
Executed onAssignorAssigneeConveyanceFrameReelDoc
Jun 06 1994KORMOCZY, PETERChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994TOROK, ZOLTANChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994SZATMARI, ISTVANChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994HERMECZ, ISTVANChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994MARMAROSI, KATALINChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994SZABO, ANNA Z Chinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994LENGYEL, JOZSEFChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994SZIRAKI, ISTVANChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994GAAL, JOZSEFChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 06 1994MAGYAR, KALMANChinoin Gyogyszer es Vegyeszeti Termekek Gyara RtASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS 0071310233 pdf
Jun 17 1994Chinoin Gyogyszer es Vegyeszeti Termekek Gyara Rt.(assignment on the face of the patent)
Date Maintenance Fee Events
Jan 07 1998ASPN: Payor Number Assigned.
Jul 25 2000REM: Maintenance Fee Reminder Mailed.
Dec 31 2000EXP: Patent Expired for Failure to Pay Maintenance Fees.


Date Maintenance Schedule
Dec 31 19994 years fee payment window open
Jul 01 20006 months grace period start (w surcharge)
Dec 31 2000patent expiry (for year 4)
Dec 31 20022 years to revive unintentionally abandoned end. (for year 4)
Dec 31 20038 years fee payment window open
Jul 01 20046 months grace period start (w surcharge)
Dec 31 2004patent expiry (for year 8)
Dec 31 20062 years to revive unintentionally abandoned end. (for year 8)
Dec 31 200712 years fee payment window open
Jul 01 20086 months grace period start (w surcharge)
Dec 31 2008patent expiry (for year 12)
Dec 31 20102 years to revive unintentionally abandoned end. (for year 12)